1
|
Bagaev A, Pichugin A, Nelson EL, Agadjanyan MG, Ghochikyan A, Ataullakhanov RI. Anticancer Mechanisms in Two Murine Bone Marrow-Derived Dendritic Cell Subsets Activated with TLR4 Agonists. THE JOURNAL OF IMMUNOLOGY 2018; 200:2656-2669. [PMID: 29500244 DOI: 10.4049/jimmunol.1701126] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 02/05/2018] [Indexed: 12/24/2022]
Abstract
Dendritic cells (DCs) are well-known for their functions in orchestrating the innate and adaptive arms of immune defense. However, under certain conditions, DCs can exert tumoricidal activity. We have elucidated the mechanism of tumor suppression by TLR4-activated bone marrow-derived DCs (BMDCs) isolated from BALB/c mice. We identified that two distinct subsets of BMDCs (CD11b+CD11c+I-A/Eint and CD11b+CD11c+I-A/Ehigh) have different cytotoxic mechanisms of action. The cytotoxicity of the former subset is mediated through NO and reactive oxygen species and type I IFN (IFN-β), whereas the latter subset acts only through IFN-β. TLR4 agonists, LPS or pharmaceutical-grade ImmunoMax, activate CD11c+ BMDCs, which, in turn, directly kill 4T1 mouse breast cancer cells or inhibit their proliferation in an MHC-independent manner. These data define two populations of BMDCs with different mechanisms of direct cytotoxicity, as well as suggest that the I-A/Eint subset could be less susceptible to counteracting mechanisms in the tumor microenvironment and support investigation of similar subsets in human DCs.
Collapse
Affiliation(s)
- Alexander Bagaev
- The Institute of Immunology, Federal Medical-Biological Agency, Moscow 115478, Russia
| | - Aleksey Pichugin
- The Institute of Immunology, Federal Medical-Biological Agency, Moscow 115478, Russia
| | - Edward L Nelson
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA 92697.,Division of Hematology and Oncology, Department of Medicine, University of California, Irvine, Irvine, CA 92697.,Chao Family Comprehensive Cancer Center, University of California, Irvine, Irvine, CA 92868
| | - Michael G Agadjanyan
- Department of Molecular Immunology, Institute for Molecular Medicine, Huntington Beach, CA 92647; and.,The Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA 92697
| | - Anahit Ghochikyan
- Department of Molecular Immunology, Institute for Molecular Medicine, Huntington Beach, CA 92647; and
| | | |
Collapse
|
2
|
Shimanovsky A, Jethava A, Dasanu CA. Immune alterations in malignant melanoma and current immunotherapy concepts. Expert Opin Biol Ther 2013; 13:1413-27. [PMID: 23930800 DOI: 10.1517/14712598.2013.827658] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
INTRODUCTION Malignant melanoma is a highly aggressive, immunogenic tumor that has the ability to modulate the immune system to its own advantage. Patients with melanoma present numerous cellular immune defects and cytokine abnormalities, all leading to suppression of the host anti-tumor immune response. Innovative treatment strategies can be achieved through employing our knowledge of the melanoma-induced immune alterations. AREAS COVERED The authors review comprehensively the immune abnormalities in individuals with melanoma, and provide a summary of currently available melanoma immunotherapy agents that are currently on the market or undergoing clinical trials. EXPERT OPINION Ipilimumab, a monoclonal antibody directed against the CTLA-4, is one of the current forefront treatment strategies in malignant melanoma. Novel immunomodulating agents have shown clear activity in patients with malignant melanoma. These include anti-PD-1 and anti-PD-1 ligand antibodies that may soon become important items in the anti-melanoma armamentarium. Combinations of different immunotherapy agents, between themselves or with other agents, are currently being studied in an attempt to further enhance the antineoplastic effect in patients with malignant melanoma.
Collapse
Affiliation(s)
- Alexei Shimanovsky
- University of Connecticut Health Science Center, Department of Medicine , Farmington, 21 Temple Street # 501, Hartford, CT 06103 , USA
| | | | | |
Collapse
|
3
|
Maslin B, Alexandrescu DT, Ichim TE, Dasanu CA. Newer developments in the immunotherapy of malignant melanoma. J Oncol Pharm Pract 2013; 20:3-10. [DOI: 10.1177/1078155212472702] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Individuals with malignant melanoma present a variety of immune abnormalities including but not limited to cellular immune dysfunction, antigen presentation deficits, and cytokine production defects. Therefore, enhancing the immune system potential represents an appealing avenue for melanoma therapy. The authors review the immune therapies currently in clinical use as well as the most promising immunotherapy candidates. Ipilimumab, a monoclonal antibody against the CTLA-4, was approved for the therapy of advanced melanoma in 2011. In addition, sizeable anti-melanoma activity has recently been shown with the use of other agents including anti-PD-1/anti-PD-1 ligand antibodies. Consequently, these experimental immunotherapy agents may soon become important items in the anti-melanoma armamentarium.
Collapse
Affiliation(s)
- Benjamin Maslin
- Department of Medicine, Yale University School of Medicine, New Haven, CT, USA
| | | | | | - Constantin A Dasanu
- Department of Hematology-Oncology, St. Francis Hospital and Medical Center, Hartford, CT, USA
| |
Collapse
|
4
|
Alexandrescu DT, Ichim TE, Riordan NH, Marincola FM, Di Nardo A, Kabigting FD, Dasanu CA. Immunotherapy for melanoma: current status and perspectives. J Immunother 2010; 33:570-90. [PMID: 20551839 PMCID: PMC3517185 DOI: 10.1097/cji.0b013e3181e032e8] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Immunotherapy is an important modality in the therapy of patients with malignant melanoma. As our knowledge about this disease continues to expand, so does the immunotherapeutic armamentarium. Nevertheless, successful preclinical models do not always translate into clinically meaningful results. The authors give a comprehensive analysis of most recent advances in the immune anti-melanoma therapy, including interleukins, interferons, other cytokines, adoptive immunotherapy, biochemotherapy, as well as the use of different vaccines. We also present the fundamental concepts behind various immune enhancement strategies, passive immunotherapy, as well as the use of immune adjuvants. This review brings into discussion the results of newer and older clinical trials, as well as potential limitations and drawbacks seen with the utilization of various immune therapies in malignant melanoma. Development of novel therapeutic approaches, along with optimization of existing therapies, continues to hold a great promise in the field of melanoma therapy research. Use of anti-CTLA4 and anti-PD1 antibodies, realization of the importance of co-stimulatory signals, which translated into the use of agonist CD40 monoclonal antibodies, as well as activation of innate immunity through enhanced expression of co-stimulatory molecules on the surface of dendritic cells by TLR agonists are only a few items on the list of recent advances in the treatment of melanoma. The need to engineer better immune interactions and to boost positive feedback loops appear crucial for the future of melanoma therapy, which ultimately resides in our understanding of the complexity of immune responses in this disease.
Collapse
Affiliation(s)
- Doru T Alexandrescu
- Division of Dermatology, University of California at San Diego, San Diego, CA, USA.
| | | | | | | | | | | | | |
Collapse
|
5
|
Minuzzo S, Moserle L, Indraccolo S, Amadori A. Angiogenesis meets immunology: Cytokine gene therapy of cancer. Mol Aspects Med 2007; 28:59-86. [PMID: 17306360 DOI: 10.1016/j.mam.2006.12.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2006] [Accepted: 12/29/2006] [Indexed: 01/19/2023]
Abstract
Delivery of cytokine genes at the tumor site in pre-clinical models has been shown to recruit host inflammatory cells followed by inhibition of tumor growth. This local effect is often accompanied by systemic protection mediated by the immune system, mainly by CD8(+) T and NK cells. On this basis, cytokine gene-transduced tumor cells have widely been used as vaccines in clinical trials, which have shown good safety profiles and some local responses but substantial lack of systemic efficacy. Are these findings the end of the story? Possibly not, if major improvements will be attained in the coming years. These should be directed at the level of gene selection and delivery, in order to identify the optimal cytokine and achieve efficient and durable cytokine expression, and at the level of improving immune stimulation, i.e. by co-administration of co-stimulatory molecules including B7 and CD40, or boosting the expression of tumor antigens or MHC class I molecules. Interestingly, some of the cytokines which have shown encouraging anti-tumor activity, including IFNs, IL-4, IL-12 and TNF-alpha, are endowed with anti-angiogenic or vasculotoxic effects, which may significantly contribute to local tumor control. Therapeutic exploitation of this property may result in the design of novel approaches which, by maximizing immune-stimulating and anti-angiogenic effects, could possibly lead to starvation of established tumors in patients.
Collapse
Affiliation(s)
- Sonia Minuzzo
- Department of Oncology and Surgical Sciences, University of Padova, via Gattamelata 64, 35128 Padova, Italy
| | | | | | | |
Collapse
|
6
|
Brown L, Roda J, Terrell C, Chaudhury AR, Crespin T, Carson WE, Lesinski GB. Interferon alpha and CPG oligodeoxynucleotides elicit additive immunostimulatory and antitumor effects. Surgery 2006; 140:297-306. [PMID: 16904983 DOI: 10.1016/j.surg.2006.05.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2006] [Revised: 04/27/2006] [Accepted: 05/06/2006] [Indexed: 11/28/2022]
Abstract
BACKGROUND We hypothesized that interferon alpha (IFN-alpha) and unmethylated cytosine-phosphothioate-guanine (CpG)-rich oligodexoynucleotides (CpG ODNs) would elicit potent antitumor activity due to the ability of this treatment combination to activate complimentary signal transduction intermediates. METHODS Peripheral blood mononuclear cells treated with CpG ODNs, IFN-alpha, or both agents combined were evaluated for cytotoxicity against human melanoma cells, Jak-STAT signal transduction by flow cytometry, and ISG-15 gene expression by real-time polymerase chain reaction. The effects of CpG ODNs and IFN-alpha were evaluated in murine models of melanoma in wild-type, IFN-gamma-deficient, and STAT1-deficient mice. Negative controls in all experiments included treatment with control ODN or phosphate-buffered saline. RESULTS Treatment of peripheral blood mononuclear cells with a combination of CpG ODNs and IFN-alpha resulted in enhanced cytotoxicity, activation of natural killer cells, IFN-alpha-induced STAT1 phosphorylation, and transcription levels of ISG-15. These immunostimulatory effects of CpG ODNs were associated with increased expression of STAT1 and STAT2 proteins. Administration of CpG ODNs plus IFN-alpha elicited superior antitumor activity in a murine model of B16 melanoma compared with either agent alone. The antitumor properties of CpG ODNs were dependent on STAT1-mediated signal transduction within the host but independent of endogenously produced IFN-gamma. CONCLUSIONS CpG ODNs represent potent immune stimulants that elicit antitumor effects through STAT1-mediated signal transduction.
Collapse
Affiliation(s)
- Lloyd Brown
- Department of Surgery, Human Cancer Genetics Program, Columbus, OH, USA
| | | | | | | | | | | | | |
Collapse
|
7
|
Olson MV, Lee J, Zhang F, Wang A, Dong Z. Inducible nitric oxide synthase activity is essential for inhibition of prostatic tumor growth by interferon-beta gene therapy. Cancer Gene Ther 2006; 13:676-85. [PMID: 16470211 DOI: 10.1038/sj.cgt.7700941] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
We have previously reported that adenoviral vector-mediated interferon (IFN)-beta gene therapy inhibits orthotopic growth of human prostate cancer cells in nude mice. The purpose of this study was to determine efficacy and mechanisms of this therapy in immune-competent mice. TRAMP-C2Re3 mouse prostate cancer cells infected with 100 multiplicity of infection (MOI) of adenoviral vector encoding for mouse IFN-beta (AdmIFN-beta), but not AdE/1 (a control adenoviral vector), produced approximately 60 ng/10(5) cells/24 h of IFN-beta. The tumorigenicity of AdmIFN-beta-transduced cells was dramatically reduced in the prostates of C57BL/6 mice. A single intratumoral injection of 2 x 10(9) PFU (plaque-forming unit) of AdmIFN-beta inhibited tumor growth by 70% and prolonged survival of tumor-bearing mice. Intriguingly, this AdmIFN-beta therapy did not alter the growth of tumors in inducible nitric oxide synthase (iNOS)-null C57BL/6 mice. Immunohistochemical analysis revealed that treatment of tumors with AdmIFN-beta in wild-type C57BL/6 mice led to increased iNOS expression, decreased microvessel density, decreased cell proliferation, and increased apoptosis. Furthermore, quantitative reverse-transcriptional PCR analysis showed that AdmIFN-beta therapy, in C57BL/6 but not the iNOS-null counterparts, reduced levels of the mRNAs for angiopoietin, basic fibroblast growth factor, matrix metalloproteinase-9, transforming growth factor-beta1, vascular endothelial growth factor (VEGF)-A, and VEGF-B, as well as the antiapoptotic molecule endothelin-1. These data indicated that IFN-beta gene therapy could be effective alternative for the treatment of locally advanced prostate cancer and suggest an obligatory role of NO in IFN-beta antitumoral effects in vivo.
Collapse
Affiliation(s)
- M V Olson
- Department of Neuro-Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | | | | | | |
Collapse
|
8
|
Lesinski GB, Badgwell B, Zimmerer J, Crespin T, Hu Y, Abood G, Carson WE. IL-12 pretreatments enhance IFN-alpha-induced Janus kinase-STAT signaling and potentiate the antitumor effects of IFN-alpha in a murine model of malignant melanoma. THE JOURNAL OF IMMUNOLOGY 2004; 172:7368-76. [PMID: 15187113 DOI: 10.4049/jimmunol.172.12.7368] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
IFN-alpha 2b (IFN-alpha) has been used to treat patients with metastatic malignant melanoma and patients rendered disease-free via surgery but at high risk for recurrence. We hypothesized that IL-12 pretreatments would result in endogenous IFN-gamma production, and that this, in turn, would up-regulate levels of Janus kinase-STAT signaling intermediates and lead to increased expression of genes regulated by IFN-alpha. Treatment of PBMCs with IL-12 stimulated a significant and dose-dependent production of IFN-gamma. Pretreatment of PBMCs and tumor cells with IFN-gamma-containing supernatants from IL-12-stimulated PBMCs led to up-regulation of STAT1, STAT2, and IFN regulatory factor 9 (IRF9) and potentiated IFN-alpha-induced STAT signaling within PBMCs and tumor cells. These effects were abrogated by neutralization of IFN-gamma in the PBMC supernatants with an anti-IFN-gamma Ab. Pretreatment of HT144 melanoma cells and PBMCs with IFN-gamma or IFN-gamma-containing supernatants enhanced the actions of IFN-alpha at the transcriptional level, as measured by real-time RT PCR analysis of the IFN-stimulated gene 15. Experiments in wild-type C57BL/6 and IFN-gamma receptor knockout (B6.129S7-Ifngr(tm1Agt)) mice demonstrated that a regimen of IL-12 pretreatment, followed by IFN-alpha, could cure mice of i.p. B16F1 melanoma tumors (p < 0.007), whereas mice treated with either agent alone or PBS succumbed to fatal tumor burden. However, this treatment regimen did not significantly prolong the survival of IFN-gamma-deficient (B6.129S7-Ifng(tm1Ts)) mice compared with mice treated with IFN-alpha alone. These results suggest that the response to IFN-alpha immunotherapy can be significantly enhanced by IL-12 pretreatment, and this effect is dependent upon endogenous IFN-gamma production and its actions on melanoma cells.
Collapse
Affiliation(s)
- Gregory B Lesinski
- Department of Human Cancer Genetics, Ohio State University, Columbus, OH 43210, USA
| | | | | | | | | | | | | |
Collapse
|
9
|
Badgwell B, Lesinski GB, Magro C, Abood G, Skaf A, Carson W. The antitumor effects of interferon-alpha are maintained in mice challenged with a STAT1-deficient murine melanoma cell line. J Surg Res 2004; 116:129-36. [PMID: 14732359 DOI: 10.1016/j.jss.2003.09.005] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
INTRODUCTION Interferon-alpha (IFN-alpha) is currently administered to patients with metastatic malignant melanoma and those who are at risk for recurrence following surgery for high-risk lesions. Signal transducer and activator of transcription 1 (STAT1) is a transcription factor that is activated by IFN-alpha and is thought to mediate the majority of its antitumor effects. Loss of STAT1 has been found in IFN-resistant melanoma cells. We developed a murine melanoma cell line in a STAT1-deficient mouse. We also transfected B16 melanoma cells with a wild-type form of STAT1 to induce its overexpression. Using the resulting cell lines and STAT1-deficient mice, we tested whether IFN-alpha could exert an antitumor effect on melanoma cells in the absence of STAT1-mediated signal transduction. MATERIALS AND METHODS A melanoma tumor was induced in STAT1-deficient mice via the application of DMBA (tumor initiator) followed by croton oil (tumor promoter). Immunohistochemical analysis confirmed that the resulting tumor was a malignant melanoma. Immunoblot analysis, intracellular flow cytometry, and gel-shift analysis were used to confirm the lack of STAT1 in the derivative cell line (AGS-1). In addition, the STAT1 protein was overexpressed in B16 melanoma cells by stable transfection with a plasmid construct encoding wild-type STAT1. The effects of IFN-alpha on these cell lines were studied in vitro and in vivo. RESULTS STAT1 was not expressed in the AGS-1 murine melanoma cell line. Treatment with IFN-alpha did not lead to activation of STAT1. Cell proliferation assays revealed that while IFN-alpha did not exert an antiproliferative effect on this cell line, it was capable of prolonging the survival of STAT1-competent C57BL/6 mice bearing 1 x 10(6) AGS-1 tumor cells in the intraperitoneal position (n = 20, P < 0.05), as compared to PBS-treated controls. Also, the survival of IFN-alpha-treated mice (as compared to PBS-treated controls) was not affected by the overexpression of STAT1 in B16 tumor cells. CONCLUSIONS This data suggests that IFN-alpha can enhance survival in an animal model where STAT1-mediated signal transduction and gene regulation is absent within the tumor but is present within the host. This data also indicates that the overexpression of STAT1 within the tumor does not significantly enhance the effects of exogenously administered IFN-alpha in this model. These findings indicate that the bulk of the antitumor actions of IFN-alpha may be derived from its effects on host tissues.
Collapse
Affiliation(s)
- Brian Badgwell
- Department of Surgery, Division of Surgical Oncology, Arthur G. James Cancer Hospital and Richard B. Solove Research Institute, The Ohio State University, Columbus, Ohio 43210, USA
| | | | | | | | | | | |
Collapse
|
10
|
Lesinski GB, Anghelina M, Zimmerer J, Bakalakos T, Badgwell B, Parihar R, Hu Y, Becknell B, Abood G, Chaudhury AR, Magro C, Durbin J, Carson WE. The antitumor effects of IFN-alpha are abrogated in a STAT1-deficient mouse. J Clin Invest 2003; 112:170-80. [PMID: 12865406 PMCID: PMC164283 DOI: 10.1172/jci16603] [Citation(s) in RCA: 88] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
IFN-alpha activates the signal transducer and activator of transcription (STAT) family of proteins; however, it is unknown whether IFN-alpha exerts its antitumor actions primarily through a direct effect on malignant cells or by stimulating the immune system. To investigate the contribution of STAT1 signaling within the tumor, we generated a STAT1-deficient melanoma cell line, AGS-1. We reconstituted STAT1 into AGS-1 cells by retroviral gene transfer. The resulting cell line (AGS-1STAT1) showed normal regulation of IFN-alpha-stimulated genes (e.g., H2k, ISG-54) as compared with AGS-1 cells infected with the empty vector (AGS-1MSCV). However, mice challenged with the AGS-1, AGS-1STAT1, and AGS-1MSCV cell lines exhibited nearly identical survival in response to IFN-alpha treatment, indicating that restored STAT1 signaling within the tumor did not augment the antitumor activity of IFN-alpha. In contrast, STAT1-/- mice could not utilize exogenous IFN-alpha to inhibit the growth of STAT1+/+ melanoma cells in either an intraperitoneal tumor model or in the adjuvant setting. The survival of tumor-bearing STAT1-/- mice was identical regardless of treatment (IFN-alpha or PBS). Additional cell depletion studies demonstrated that NK cells mediated the antitumor effects of IFN-alpha. Thus, STAT1-mediated gene regulation within immune effectors was necessary for mediating the antitumor effects of IFN-alpha in this experimental system.
Collapse
Affiliation(s)
- Gregory B Lesinski
- Department of Human Cancer Genetics, The Ohio State University, Columbus, Ohio 43210, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Chiang EY, Henson M, Stroynowski I. Correction of defects responsible for impaired Qa-2 class Ib MHC expression on melanoma cells protects mice from tumor growth. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2003; 170:4515-23. [PMID: 12707328 DOI: 10.4049/jimmunol.170.9.4515] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
One of the principal mechanisms of tumor immune evasion is alteration of class I MHC expression. We have identified defects contributing to down-regulation of class I MHC expression in the widely studied murine B16 melanoma and its variants B16F1, B16F10, BL6-2, BL6-8 and B78H1. Transcription of the nonclassical class I MHC genes Q8 and Q9 (Qa-2 Ags) has been switched off in the entire panel of melanoma lines, suggesting that this event occurred early during tumor progression. B78H1, unlike B16F1 and B16F10 sublines, is also selectively devoid of TAP2 and low molecular weight protein 7 as well as classical class I MHC K(b) and D(b) transcripts. Cotransfection of B78H1 with TAP2 and class I H chain genes is sufficient to reconstitute surface expression of exogenously delivered class I MHC without concomitant re-expression of endogenous beta(2)-microglobulin-associated class I. The serological absence of endogenous class Ia and Ib at the surface of TAP2-negative as well as TAP2-transfected B78H1 makes this system a suitable model for studying the properties of isolated class I proteins in tumors. We used this system to demonstrate that B78H1 cells genetically manipulated to re-express Q9 Ag have reduced tumor potential in syngeneic B6 mice compared with TAP2-transfected parental melanoma. Both NK cells and CTLs appear to collaborate in restraining growth of Q9-positive tumors. The results implicate Qa-2 in antitumor responses and illustrate the utility of the B78H1 system for identifying in vivo interactions between class I MHC molecules of interest and immune cells of innate and/or adaptive immunity.
Collapse
MESH Headings
- ATP Binding Cassette Transporter, Subfamily B, Member 3
- ATP-Binding Cassette Transporters/genetics
- Animals
- Antigen Presentation/genetics
- CD8-Positive T-Lymphocytes/immunology
- Cell Division/genetics
- Cell Division/immunology
- Cell Line, Transformed
- Down-Regulation/genetics
- Down-Regulation/immunology
- Genetic Vectors
- Granulocyte-Macrophage Colony-Stimulating Factor/genetics
- Histocompatibility Antigens Class I/biosynthesis
- Histocompatibility Antigens Class I/genetics
- Immunologic Deficiency Syndromes/genetics
- Immunologic Deficiency Syndromes/immunology
- Killer Cells, Natural/immunology
- Melanoma, Experimental/immunology
- Melanoma, Experimental/pathology
- Melanoma, Experimental/prevention & control
- Mice
- Mice, Inbred C57BL
- Neoplasm Transplantation
- Transduction, Genetic/methods
- Transfection
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- Eugene Y Chiang
- Center for Immunology, Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | | | | |
Collapse
|
12
|
Lu W, Dong Z, Donawho C, Fidler IJ. Specific immunotherapy against occult cancer metastases. Int J Cancer 2002; 100:480-5. [PMID: 12115534 DOI: 10.1002/ijc.10494] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
We investigated the efficacy of a preparation containing High Five (H5) insect cells infected with recombinant baculovirus encoding the murine interferon-beta gene (H5BVIFN-beta) against established primary tumors and occult lung metastases. Injection of live or lyophilized H5BVIFN-beta into established subcutaneous tumors of the highly metastatic murine UV-2237m fibrosarcoma or K-1735M2 melanoma in syngeneic mice eradicated both primary tumors and preexisting lung metastases. The therapeutic effects of H5BVIFN-beta were not observed in nude mice and were diminished in syngeneic mice depleted of CD4(+) and CD8(+) T cells. Immunohistochemical staining showed that tumors injected with H5BVIFN-beta were densely infiltrated by CD4(+) and CD8(+) T cells in mice with normal CD4/CD8 complement. These data demonstrate that, unlike most immunologic approaches in which prophylaxis can be achieved but eradication of established tumor is rare, lyophilized preparations of H5BVIFN-beta can serve as a novel immunotherapy against both primary tumors and their occult metastases.
Collapse
Affiliation(s)
- Weixin Lu
- Department of Cancer Biology, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA
| | | | | | | |
Collapse
|
13
|
Wu TY, Fleischmann WR. Murine B16 melanoma vaccination-induced tumor immunity: identification of specific immune cells and functions involved. J Interferon Cytokine Res 2001; 21:1117-27. [PMID: 11798470 DOI: 10.1089/107999001317205259] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022] Open
Abstract
Vaccination using inactivated B16 melanoma cells that have been treated in vitro for > 2 weeks with interferon-alpha (IFN-alpha) (B16alpha cells) has been shown to elicit a protective host antitumor immunity. In these studies, vaccination with B16alpha cells has been shown to provide protection against primary B16 tumor challenge, established B16 tumors, and metastatic B16 tumors. Specific immune cells and factors that might mediate this tumor immunity have now been evaluated. Macrophage depletion studies suggest that macrophage function is required for expression of tumor immunity either for processing of antigen or for cytokine production but that macrophage function is not involved in direct cytotoxicity against the B16 challenge tumor. CD8(+) T cell depletion studies show that cytotoxic T cell function is required for expression of tumor immunity. Syngeneic knockout mouse experiments offer further insights into the immune cells and factors that mediate the development and expression of tumor immunity. First, interleukin-12 (IL-12) knockout mouse experiments identify IL-12 as an important cytokine in mediating the development of tumor immunity. Second, specific knockout mouse experiments show that tumor immunity requires the function of CD4(+) T cells, CD8(+) T cells, and natural killer (NK) cells. Third, specific knockout mouse experiments show that tumor immunity does not require the function of B cells. The results suggest that vaccination with inactivated B16alpha cells induces an active, cell-mediated immunity to B16 melanoma cells. The tumor vaccination protocol with B16alpha cell vaccinations establishes a potent tumor immunity against B16 melanoma tumors in mice and may serve as a model for induction of tumor immunity against primary or secondary melanoma tumors in humans.
Collapse
Affiliation(s)
- T Y Wu
- Department of Microbiology and Immunology, The University of Texas Medical Branch, Galveston, TX 77555, USA
| | | |
Collapse
|
14
|
Goł b J, Zagozdzon R, Kamiński R, Kozar K, Gryska K, Izycki D, Mackiewicz A, Stokłosa T, Giermasz A, Lasek W, Jakóbisiak M. Potentiatied antitumor effectiveness of combined chemo-immunotherapy with interleukin-12 and 5-fluorouracil of L1210 leukemia in vivo. Leukemia 2001; 15:613-20. [PMID: 11368364 DOI: 10.1038/sj.leu.2402076] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
In this study we investigated the efficacy of a combination of IL-12 and 5-FU, a chemotherapeutic exerting several immunomodulatory effects, in murine L1210 leukemia. Mice inoculated with 1 x 10(5) leukemia cells were treated with a single dose of 5-FU (50 mg/kg) and seven daily doses of IL-12 (100 ng/dose), and were observed for survival. Treatment with IL-12 or 5-FU given alone produced moderate anti-leukemic effects. However, combination of both drugs resulted in a significant prolongation of mouse survival time. Importantly, there were 70% of long-term (>60 days) survivors among mice treated with both agents simultaneously. Moreover, we observed 100% of long-term survivors when mice were treated with a minimally increased dose of IL-12 (170 ng) in combination with 5-FU (50 mg/kg). The antileukemic effects were completely abrogated in scid/scid mice and in mice depleted of peritoneal macrophages and significantly decreased after administration of anti-CD3+, anti-CD4+ or anti-CD8+ monoclonal antibodies. Administration of anti-NK1.1 antibodies did not decrease the antileukemic effects indicating that NK cells are not important effectors of this treatment regimen. Collectively, these results indicate that the combination of IL-12 and 5-FU is inducing strong antileukemic responses that are dependent on the presence and activity of macrophages and T lymphocytes and warrant further studies of combined chemo-immunotherapy with IL-12.
Collapse
Affiliation(s)
- J Goł b
- Department of Immunology, Center of Biostructsure Research, Medical University of Warsaw, Poland
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Abstract
Vaccination with a tumour antigen-expressing plasmid DNA (pDNA) is a novel approach to human cancer immunotherapy. Initial results in preclinical rodent tumour models are promising, revealing that pDNA cancer vaccines can elicit both humoral, as well as cell-mediated immunity and, in some cases, protect against tumour growth. Compared to peptide, viral or dendritic cell vaccines, the delivery of tumour antigens using pDNA has the advantages of ease of manufacture, lack of toxicity and broad applicability to large populations. With advances in modern genomics strategies and the identification of an increasing number of tumour antigen genes, pDNA-based cancer vaccines may be used in the future to treat a wide variety of human cancers.
Collapse
|
16
|
Wu TY, Fleischmann WR. Efficacy of B16 melanoma cells exposed in vitro to long-term IFN-alpha treatment (B16alpha cells) as a tumor vaccine in mice. J Interferon Cytokine Res 1998; 18:829-39. [PMID: 9809618 DOI: 10.1089/jir.1998.18.829] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
B16 melanoma cells exposed to >2 weeks of in vitro interferon-alpha (IFN-alpha) treatment (B16alpha cells) were UV inactivated and used for vaccination. This vaccination was efficacious against challenge with parental B16 cells in the absence of adjuvant therapy. Vaccinations based on parental cells and B16 cells exposed to short-term in vitro IFN-alpha treatment were not effective. The efficacy of B16alpha vaccination was evaluated using three B16 tumor models. Using intraperitoneal (i.p.) tumor challenge given after vaccination, vaccination efficacy depended on the concentration of IFN-alpha to which B16alpha cells were exposed, the number of inactivated B16alpha cells inoculated, the number of inoculations administered, and the amount of tumor burden. A significant fraction (30%) of vaccinated mice surviving initial challenge had durable immunity against a second parental tumor challenge. This immunity increased to 92% with administration of a single booster vaccination. Using metastatic tumor challenge given after vaccination, vaccination reduced lung metastases by approximately 67%. Using vaccination begun 3 days after subcutaneous (s.c.) tumor challenge, regression of established tumor occurred when vaccination was given i.p. (39%) or contralaterally s.c. (53%). Taken together, the results suggest that vaccination with inactivated B16alpha cells may serve as a model for induction of host tumor immunity against primary or secondary tumors.
Collapse
Affiliation(s)
- T Y Wu
- Department of Microbiology and Immunology, The University of Texas Medical Branch, Galveston 77555, USA
| | | |
Collapse
|
17
|
Zagozdzon R, Gołab J, Stokłosa T, Giermasz A, Nowicka D, Feleszko W, Lasek W, Jakóbisiak M. Effective chemo-immunotherapy of L1210 leukemia in vivo using interleukin-12 combined with doxorubicin but not with cyclophosphamide, paclitaxel or cisplatin. Int J Cancer 1998; 77:720-7. [PMID: 9688305 DOI: 10.1002/(sici)1097-0215(19980831)77:5<720::aid-ijc10>3.0.co;2-5] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
It has been well established that chemo-immunotherapy using cytotoxic drugs and appropriate cytokines offers a new approach to increasing the therapeutic index in the treatment of neoplastic diseases. This study investigates the efficacy of combinations of interleukin-12 with cyclophosphamide, paclitaxel, cisplatin or doxorubicin in the murine L1210 leukemia model. Mice inoculated i.p. with 1 x 10(3) or 1 x 10(5) leukemia cells were treated with interleukin-12 and/or chemotherapeutics, and were observed daily for survival. Immunosuppression with X-irradiation or macrophage depletion with injections of silica were used to examine the dependence of the therapeutic effects on the efficiency of the immune system. Treatment with interleukin-12 or one of the studied chemotherapeutics given alone resulted in moderate antileukemic effects. Combination of interleukin-12 with cyclophosphamide or paclitaxel produced no augmentation of anti-leukemic effects in comparison with these agents given alone. Combination of interleukin-12 with cisplatin resulted in prolongation of the survival time; however, in the experiment with mice inoculated with 1 x 10(5) leukemia cells, no long-term survivors (>60 days) were observed; on the contrary, combination of interleukin-12 with doxorubicin resulted in 100% long-term survivors. This effect was completely abrogated either by X-irradiation of mice or by macrophage depletion. We also found that doxorubicin augments IL-12-stimulated production of interferon-gamma in vivo. Our observations demonstrating potentiation of the antileukemic effects of the IL-12 and doxorubicin combination suggest that the combined use of these 2 agents could be beneficial in leukemia therapy.
Collapse
Affiliation(s)
- R Zagozdzon
- Department of Immunology, Institute of Biostructure, Medical University of Warsaw, Poland.
| | | | | | | | | | | | | | | |
Collapse
|
18
|
Böhm W, Thoma S, Leithäuser F, Möller P, Schirmbeck R, Reimann J. T Cell-Mediated, IFN-γ-Facilitated Rejection of Murine B16 Melanomas. THE JOURNAL OF IMMUNOLOGY 1998. [DOI: 10.4049/jimmunol.161.2.897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Abstract
The murine melanoma cell line B16.F10 (H-2b) was used to study specific T cell responses that reject tumors. Stable B16 transfectants were established that express viral Ags, either the hepatitis B surface Ag (HBsAg) or the large tumor Ag (T-Ag) of SV40. B16 cells and their transfected sublines were CD40+CD44+ but expressed no (or low levels of the) costimulator molecules CD154 (CD40L), CD48, CD54, CD80, and CD86. Surface expression of MHC class I (Kb, Db) and class II (I-Ab) molecules by B16 cells was low, but strikingly up-regulated by IFN-γ. CD95 (Fas) and CD95 ligand (CD95L (FasL)) were “spontaneously” expressed by B16 cells growing in vitro in serum-free medium; these markers were strikingly up-regulated by IFN-γ. B16 cells coexpressing CD95 and CD95L were irreversibly programed for apoptosis. In vitro, noninduced B16 transfectants stimulated a specific IFN-γ release response, but no cytolytic response (in a 4-h assay) in MHC class I-restricted CTL; in contrast, IFN-γ-induced B16 targets were efficiently and specifically lysed by CTL. In vivo, B16 transfectants were specifically rejected by DNA-vaccinated syngeneic hosts through a T-dependent immune effector mechanism. The tumors showed evidence of massive apoptosis in vivo during the rejection process. The data suggest that CTL-derived IFN-γ enhances an intrinsic suicide mechanism of these tumor cells in addition to facilitating lytic interactions of effectors with tumor targets.
Collapse
Affiliation(s)
| | | | | | - Peter Möller
- †Pathology, University of Ulm, Ulm, Germany
- Institutes of
| | | | | |
Collapse
|
19
|
Fleischmann CM, Wu TY, Fleischmann WR. B16 melanoma cells exposed in vitro to long-term IFN-alpha treatment (B16 alpha cells) as activators of tumor immunity in mice. J Interferon Cytokine Res 1997; 17:37-43. [PMID: 9041470 DOI: 10.1089/jir.1997.17.37] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Mice inoculated with B16 melanoma cells exposed to long-term in vitro IFN-alpha treatment (> or = 14 days, B16 alpha cells) but not short-term in vitro IFN-alpha treatment (24 h) exhibited an enhanced survival time. Enhanced survival time also occurred when inactivated B16 alpha cells were inoculated at the same time as live B16 cells. Further, an even greater improvement in survival time was observed when the inactivated B16 alpha cells were inoculated before live B16 cell challenge. No enhancement in survival time was observed when mice were inoculated with inactivated, untreated B16 cells. Enhancement of survival time by B16 alpha cells was unrelated to retrovirus surface antigen expression. Long-lasting protective immunity to B16 cells was observed in mice that survived B16 alpha cell, but not normal B16 cell, challenge and subsequent IFN treatment. It is evident that inoculation with inactivated B16 alpha cells, but not with inactivated untreated B16 cells, was able to prolong significantly the survival time of mice either simultaneously or subsequently challenged with live B16 cells. Additionally, survival of B16 alpha-inoculated but not B16-inoculated mice was accompanied by a durable immunity. Inoculation of inactivated B16 alpha cells may serve as a model for the induction of host immunity to a parental primary or secondary tumor.
Collapse
Affiliation(s)
- C M Fleischmann
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston 77555, USA
| | | | | |
Collapse
|